Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Hyo Je Cho is active.

Publication


Featured researches published by Hyo Je Cho.


Journal of Biological Chemistry | 2009

Structural Insight Into the Heme-Based Redox Sensing by Doss from Mycobacterium Tuberculosis.

Ha Yeon Cho; Hyo Je Cho; Young-Min Kim; Jeong Il Oh; Beom Sik Kang

Mycobacterium tuberculosis is thought to undergo transformation into its non-replicating persistence state under the influence of hypoxia or nitric oxide (NO). This transformation is thought to be mediated via two sensor histidine kinases, DosS and DosT, each of which contains two GAF domains that are responsible for detecting oxygen tension. In this study we determined the crystal structures of the first GAF domain (GAF-A) of DosS, which shows an interaction with a heme. A b-type heme was embedded in a hydrophobic cavity of the GAF-A domain and was roughly perpendicular to the β-sheet of the GAF domain. The heme iron was liganded by His-149 at the proximal heme axial position. The iron, in the oxidized form, was six-coordinated with a water molecule at the distal position. Upon reduction, the iron, in ferrous form, was five-coordinated, and when the GAF domain was exposed to atmospheric O2, the ferrous form was oxidized to generate the Met form rather than a ferrous O2-bound form. Because the heme is isolated inside the GAF domain, its accessibility is restricted. However, a defined hydrogen bond network found at the heme site could accelerate the electron transferability and would explain why DosS was unable to bind O2. Flavin nucleotides were shown to reduce the heme iron of DosS while NADH was unable to do so. These results suggest that DosS is a redox sensor and detects hypoxic conditions by its reduction.


Journal of Bacteriology | 2008

O2- and NO-Sensing Mechanism through the DevSR Two-Component System in Mycobacterium smegmatis

Jin-Mok Lee; Ha Yeon Cho; Hyo Je Cho; In-Jeong Ko; Sae Woong Park; Hyung-Suk Baik; Jee-Hyun Oh; Chi-Yong Eom; Young Min Kim; Beom Sik Kang; Jeong-Il Oh

The DevS histidine kinase of Mycobacterium smegmatis contains tandem GAF domains (GAF-A and GAF-B) in its N-terminal sensory domain. The heme iron of DevS is in the ferrous state when purified and is resistant to autooxidation from a ferrous to a ferric state in the presence of O(2). The redox property of the heme and the results of sequence comparison analysis indicate that DevS of M. smegmatis is more closely related to DosT of Mycobacterium tuberculosis than DevS of M. tuberculosis. The binding of O(2) to the deoxyferrous heme led to a decrease in the autokinase activity of DevS, whereas NO binding did not. The regulation of DevS autokinase activity in response to O(2) and NO was not observed in the DevS derivatives lacking its heme, indicating that the ligand-binding state of the heme plays an important role in the regulation of DevS kinase activity. The redox state of the quinone/quinol pool of the respiratory electron transport chain appears not to be implicated in the regulation of DevS activity. Neither cyclic GMP (cGMP) nor cAMP affected DevS autokinase activity, excluding the possibility that the cyclic nucleotides serve as the effector molecules to modulate DevS kinase activity. The three-dimensional structure of the putative GAF-B domain revealed that it has a GAF folding structure without cyclic nucleotide binding capacity.


Journal of Biological Chemistry | 2008

Determination of Three-dimensional Structure and Residues of the Novel Tumor Suppressor AIMP3/p18 Required for the Interaction with ATM

Kyungjin Kim; Min Chul Park; So Jung Choi; Young Sun Oh; Eung-Chil Choi; Hyo Je Cho; Myung Hee Kim; Soo-Hyun Kim; Dong-Wook Kim; Sung-Hoon Kim; Beom Sik Kang

Although AIMP3/p18 is normally associated with the multi-tRNA synthetase complex via its specific interaction with methionyl-tRNA synthetase, it also works as a tumor suppressor by interacting with ATM, the upstream kinase of p53. To understand the molecular interactions of AIMP3 and the mechanisms involved, we determined the crystal structure of AIMP3 at 2.0-Å resolution and identified its potential sites of interaction with ATM. AIMP3 contains two distinct domains linked by a 7-amino acid (Lys57-Ser63) peptide, which contains a 310 helix. The 56-amino acid N-terminal domain consists of two helices into which three antiparallel β strands are inserted, and the 111-amino acid C-terminal domain contains a bundle of five helices (Thr64-Tyr152) followed by a coiled region (Pro153-Leu169). Structural analyses revealed homologous proteins such as yeast glutamyl-tRNA synthetase, Arc1p, EF1Bγ, and glutathione S-transferase and suggested two potential molecular binding sites. Moreover, mutations at the C-terminal putative binding site abolished the interaction between AIMP3 and ATM and the ability of AIMP3 to activate p53. Thus, this work identified the two potential molecular interaction sites of AIMP3 and determined the residues critical for its tumor-suppressive activity through the interaction with ATM.


Journal of Biological Chemistry | 2015

Assembly of Multi-tRNA Synthetase Complex via Heterotetrameric Glutathione Transferase-homology Domains

Ha Yeon Cho; Seo Jin Maeng; Hyo Je Cho; Yoon Seo Choi; Jeong Min Chung; Sangmin Lee; Jong Hyun Kim; Chi-Yong Eom; Yeon-Gil Kim; Min Guo; Hyun Suk Jung; Beom Sik Kang; Sunghoon Kim

Background: GST domains have been found in diverse proteins involved in translational systems. Results: Four GST domains from human methionyl-tRNA synthetase, glutaminyl-prolyl-tRNA synthetase, ARS-interacting multifunctional protein (AIMP) 2, and AIMP3 are complexed in an ordered fashion. Conclusion: Four components in the human multisynthetase complex are assembled through a GST domain tetrameric complex. Significance: GST domain assemblies act as scaffolds for the formation of multicomponent protein complexes. Many multicomponent protein complexes mediating diverse cellular processes are assembled through scaffolds with specialized protein interaction modules. The multi-tRNA synthetase complex (MSC), consisting of nine different aminoacyl-tRNA synthetases and three non-enzymatic factors (AIMP1–3), serves as a hub for many signaling pathways in addition to its role in protein synthesis. However, the assembly process and structural arrangement of the MSC components are not well understood. Here we show the heterotetrameric complex structure of the glutathione transferase (GST) domains shared among the four MSC components, methionyl-tRNA synthetase (MRS), glutaminyl-prolyl-tRNA synthetase (EPRS), AIMP2 and AIMP3. The MRS-AIMP3 and EPRS-AIMP2 using interface 1 are bridged via interface 2 of AIMP3 and EPRS to generate a unique linear complex of MRS-AIMP3:EPRS-AIMP2 at the molar ratio of (1:1):(1:1). Interestingly, the affinity at interface 2 of AIMP3:EPRS can be varied depending on the occupancy of interface 1, suggesting the dynamic nature of the linear GST tetramer. The four components are optimally arranged for maximal accommodation of additional domains and proteins. These characteristics suggest the GST tetramer as a unique and dynamic structural platform from which the MSC components are assembled. Considering prevalence of the GST-like domains, this tetramer can also provide a tool for the communication of the MSC with other GST-containing cellular factors.


Journal of Structural Biology | 2011

Structural and Functional Analysis of Bacterial Flavin-Containing Monooxygenase Reveals its Ping-Pong-Type Reaction Mechanism.

Hyo Je Cho; Ha Yeon Cho; Kyung Jin Kim; Myung Hee Kim; Si Wouk Kim; Beom Sik Kang

A bacterial flavin-containing monooxygenase (bFMO) catalyses the oxygenation of indole to produce indigoid compounds. In the reductive half of the indole oxygenation reaction, NADPH acts as a reducing agent, and NADP(+) remains at the active site, protecting bFMO from reoxidation. Here, the crystal structures of bFMO and bFMO in complex with NADP(+), and a mutant bFMO(Y207S), which lacks indole oxygenation activity, with and without indole are reported. The crystal structures revealed overlapping binding sites for NADP(+) and indole, suggestive of a double-displacement reaction mechanism for bFMO. In biochemical assays, indole inhibited NADPH oxidase activity, and NADPH in turn inhibited the binding of indole and decreased indoxyl production. Comparison of the structures of bFMO with and without bound NADP(+) revealed that NADPH induces conformational changes in two active site motifs. One of the motifs contained Arg-229, which participates in interactions with the phosphate group of NADPH and appears be a determinant of the preferential binding of bFMO to NADPH rather than NADH. The second motif contained Tyr-207. The mutant bFMO(Y207S) exhibited very little indoxyl producing activity; however, the NADPH oxidase activity of the mutant was higher than the wild-type enzyme. It suggests a role for Y207, in the protection of hydroperoxyFAD. We describe an indole oxygenation reaction mechanism for bFMO that involves a ping-pong-like interaction of NADPH and indole.


Journal of Biological Chemistry | 2010

Substrate Binding Mechanism of a Type I Extradiol Dioxygenase

Hyo Je Cho; Kyungsun Kim; Seo Yean Sohn; Ha Yeon Cho; Kyung Jin Kim; Myung Hee Kim; Dockyu Kim; Beom Sik Kang

A meta-cleavage pathway for the aerobic degradation of aromatic hydrocarbons is catalyzed by extradiol dioxygenases via a two-step mechanism: catechol substrate binding and dioxygen incorporation. The binding of substrate triggers the release of water, thereby opening a coordination site for molecular oxygen. The crystal structures of AkbC, a type I extradiol dioxygenase, and the enzyme substrate (3-methylcatechol) complex revealed the substrate binding process of extradiol dioxygenase. AkbC is composed of an N-domain and an active C-domain, which contains iron coordinated by a 2-His-1-carboxylate facial triad motif. The C-domain includes a β-hairpin structure and a C-terminal tail. In substrate-bound AkbC, 3-methylcatechol interacts with the iron via a single hydroxyl group, which represents an intermediate stage in the substrate binding process. Structure-based mutagenesis revealed that the C-terminal tail and β-hairpin form part of the substrate binding pocket that is responsible for substrate specificity by blocking substrate entry. Once a substrate enters the active site, these structural elements also play a role in the correct positioning of the substrate. Based on the results presented here, a putative substrate binding mechanism is proposed.


FEBS Letters | 2011

Blockage of the Channel to Heme by the E87 Side Chain in the Gaf Domain of Mycobacterium Tuberculosis Doss Confers the Unique Sensitivity of Doss to Oxygen.

Ha Yeon Cho; Hyo Je Cho; Myung Hee Kim; Beom Sik Kang

Two sensor kinases, DosS and DosT, are responsible for recognition of hypoxia in Mycobacterium tuberculosis. Both proteins are structurally similar to each other, but DosS is a redox sensor while DosT binds oxygen. The primary difference between the two proteins is the channel to the heme present in their GAF domains. DosS has a channel that is blocked by E87 while DosT has an open channel. Absorption spectra of DosS mutants with an open channel show that they bind oxygen as DosT does when they are exposed to air, while DosT G85E mutant is oxidized similarly to DosS without formation of an oxy‐ferrous form. This suggests that oxygen accessibility to heme is the primary factor governing the oxygen‐binding properties of these proteins.


Proteins | 2007

Structural insight of the role of the Hahella chejuensis HapK protein in prodigiosin biosynthesis

Hyo Je Cho; Kyung-Jin Kim; Myung Hee Kim; Beom Sik Kang

Prodigiosin is a red pigment synthesized by a restricted group of bacteria such as Streptomyces and Serratia as a second metabolite.1,2 This chemical is a member of prodiginines, natural tripyrrole pigments, which have a common pyrroly dipyrroly methane structure.3 Although the biological activity of prodiginin in the strain has not been defined yet, it has been known to have immunosuppressive, antibacterial, antifungal, antiprotozoal, and antimalarial activities.3,4 As an anticancer agent, it induces apoptosis in various kinds of cancer cells and it also has an antimetastatic effect.5–7 Therefore, it shows some promise as a new immunosuppressant and anticancer drug. A redpigmented bacterium, Hahella chejuensis was isolated from coastal marine sediment of Jeju Island, Korea.8 These bacteria have algicidal effects and can kill Cochlodinium, which causes red tide. The bacteria produced red pigments in the cell envelope, and the main red-colored part was identified as prodigiosin. Recently, a full-genome sequence of H. chejuensis revealed hap genes, a gene cluster, which is similar to the red gene cluster for the prodigiosin biosynthesis of Streptomyces coelicolor.9 Four prodiginin biosynthetic gene clusters have been reported and the prodigiosin-biosynthetic pathway has been studied at the biochemical and molecular levels with the red gene cluster from S. coelicolor and pig gene cluster of Serratia marcescens.10–13 The Hahella hap gene cluster consists of 14 genes (hapA to hapN), and the gene content and organization resembles that of the Serratia pig gene cluster.9 All functions of hap gene products are assigned, except for HapK, which is based on red gene and pig gene products. The functions of HapK and the equivalents, PigK and RedY, are still unknown.2,13 HapK is a small protein, which consists of 105 amino acid residues. A BLAST search with the HapK sequence showed only a few homologues such as PigK and RedY. The sequence identities of HapK to PigK and RedY are 43 and 52%, respectively. No similar proteins are in any other organisms. It is very unique for the prodigiosin biosynthetic bacteria and is highly conserved among them. The pigK deletion mutant of S. marcescens suggests that PigK might function in the biosynthesis of methoxy-bipyrrole carbaldehyde (MBC), as an intermediate for prodigiosin biosynthesis.13 If HapK, PigK, or RedY are not necessary for prodigosin biosynthesis, there is no need to be conserved or unique among these bacteria. In this study, we determined the crystal structure of the hapK gene product to reveal its unknown functions based on its molecular structure.


Acta Crystallographica Section F-structural Biology and Crystallization Communications | 2008

Crystallization and preliminary crystallographic analysis of the second GAF domain of DevS from Mycobacterium smegmatis.

Ha Yeon Cho; Hyo Je Cho; Young Min Kim; Jeong Il Oh; Beom Sik Kang

Mycobacterium tuberculosis is known to transform into the nonreplicating persistence state under the influence of hypoxia or nitric oxide. DevS-DevR is a two-component regulatory system that mediates the genetic response for the transformation. DevS is a histidine kinase that contains two GAF domains for sensing hypoxia or nitric oxide. The second GAF from M. smegmatis DevS was crystallized using the sitting-drop vapour-diffusion method in the presence of sodium citrate and 2-propanol as precipitants. X-ray diffraction data were collected from crystals containing selenomethionine to a maximum resolution of 2.0 A on a synchrotron beamline. The crystals belong to the hexagonal space group P6(1). The asymmetric unit contains one molecule, corresponding to a packing density of 2.5 A(3) Da(-1). The selenium substructure was determined by the single anomalous dispersion method and structure refinement is in progress.


Journal of Biological Inorganic Chemistry | 2018

NADP+-dependent cytosolic isocitrate dehydrogenase provides NADPH in the presence of cadmium due to the moderate chelating effect of glutathione.

Hyo Je Cho; Ha Yeon Cho; Jeen-Woo Park; Oh-Shin Kwon; Hyun-Shik Lee; Tae Lin Huh; Beom Sik Kang

Collaboration


Dive into the Hyo Je Cho's collaboration.

Top Co-Authors

Avatar

Beom Sik Kang

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Ha Yeon Cho

Kyungpook National University

View shared research outputs
Top Co-Authors

Avatar

Myung Hee Kim

Korea Research Institute of Bioscience and Biotechnology

View shared research outputs
Top Co-Authors

Avatar

Jeong Il Oh

Pusan National University

View shared research outputs
Top Co-Authors

Avatar

Kyung Jin Kim

Pohang University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Kyungjin Kim

Seoul National University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dong-Wook Kim

Catholic University of Korea

View shared research outputs
Researchain Logo
Decentralizing Knowledge